The LTK antibody (e.g., AF4664 from R&D Systems) is a polyclonal immunoglobulin that specifically binds to the human LTK protein (Accession # P29376, amino acids Ser10-Pro424). Its epitope recognition is optimized for Western blotting and immunohistochemistry (IHC), with reported specificity in Jurkat T-cell leukemia lysates and human brain cortex sections . The antibody undergoes affinity purification and is validated under reducing conditions, ensuring minimal cross-reactivity with structurally similar kinases like ALK .
The antibody’s specificity was confirmed through:
Knockdown experiments: LTK depletion in HepG2 cells reduced immunofluorescence signal by >80% .
Glycosidase treatments: Resistance to PNGase F and sensitivity to EndoH confirmed LTK’s ER localization .
Cross-reactivity testing: No binding to ALK in ALK-negative cell lines (e.g., HepG2) .
4.1. Role in ER Secretion
LTK regulates ER-to-Golgi trafficking by modulating Endoplasmic Reticulum Exit Sites (ERES). Knockdown or inhibition (via alectinib/crizotinib) reduced ERES count by 30–40% in HepG2 cells . This links LTK to secretion-dependent processes like antibody production in autoimmune diseases .
4.2. Ligand Discovery
FAM150A/B proteins activate LTK signaling with EC50 values of 1.96 nM (FAM150A) and 28 pM binding affinity . These ligands may mediate LTK’s role in neurogenesis and oncogenesis .
Cancer: Elevated LTK expression in acute myeloid leukemia (AML) suggests a therapeutic target .
Autoimmunity: Gain-of-function mutations in systemic lupus erythematosus (SLE) patients correlate with hyperactive LTK signaling .
The LTK antibody is critical for:
LTK (Leukocyte tyrosine kinase, also known as protein tyrosine kinase 1 or TYK1) is a glycoprotein member of the tyrosine protein kinase family, belonging to the insulin receptor subfamily of proteins . It functions as an ER-resident receptor tyrosine kinase that regulates COPII-dependent trafficking, making it the first identified ER-resident receptor tyrosine kinase with this function . LTK is particularly important in research because it represents a potential druggable proteostasis regulator . It is expressed in various tissues including lymphocytes, cerebral cortex neurons, and cardiomyocytes, where it plays a role in cellular hypertrophy .
Endogenous LTK detection requires specific antibodies and appropriate techniques. Immunofluorescence can be performed using anti-LTK antibodies, with HepG2 cells being particularly suitable due to their high LTK expression levels and minimal ALK expression (which prevents cross-reactivity issues) . For Western blotting, available antibodies may not consistently detect endogenous LTK, so validation is essential . Alternatively, overexpression of tagged LTK constructs (e.g., flag-tagged) can facilitate detection in cellular studies . When performing immunofluorescence, co-staining with ER markers such as CLIMP63 can confirm the ER localization of LTK .
LTK antibodies are used in various research applications including:
For optimal results, antibody dilutions should be determined for each specific application and experimental system .
Validating LTK antibody specificity is crucial for reliable experimental results. The most common validation approach is to perform knockdown experiments using LTK-specific siRNAs and demonstrate a reduction in signal intensity compared to control conditions . This approach has been successfully used to validate antibody specificity in immunofluorescence studies . Additionally, comparison of staining patterns with known LTK subcellular localization (primarily ER) can provide further validation . For Western blot applications, the detection of a specific band at the expected molecular weight (~70 kDa) that diminishes upon LTK knockdown confirms antibody specificity .
LTK exists in multiple isoforms with molecular weights ranging between 50 and 100 kDa in SDS-PAGE . Distinguishing between these isoforms requires:
Selection of epitope-specific antibodies: Choose antibodies targeting regions that differ between isoforms. The database indicates several isoforms including:
High-resolution gel electrophoresis: Use gradient gels (e.g., 4-20%) to achieve better separation of closely migrating isoforms.
Isoform-specific knockdown: Employ isoform-specific siRNAs to validate antibody specificity for particular variants.
Mass spectrometry validation: Following immunoprecipitation with anti-LTK antibodies, mass spectrometry can definitively identify specific isoforms present in your sample.
When using commercial antibodies, verify the immunogen sequence to determine which isoforms the antibody is likely to recognize .
Studying LTK phosphorylation requires careful experimental design:
Antibody selection: Use phospho-specific antibodies that recognize specific phosphorylated tyrosine residues in LTK. The cytoplasmic region of LTK possesses multiple phosphotyrosines that interact with downstream signaling molecules .
Phosphatase inhibitors: Include phosphatase inhibitors (e.g., sodium orthovanadate, sodium fluoride) in all lysis buffers to preserve phosphorylation states.
Stimulation protocols: LTK activation may require specific stimulation conditions. Unlike its relative ALK, LTK is not readily accessible to extracellular ligands due to its ER localization .
Sample preparation: Rapid sample processing is essential to prevent dephosphorylation. Use hot SDS sample buffer or direct lysis in Laemmli buffer for immediate denaturation of phosphatases.
Controls: Include samples treated with phosphatase to confirm the specificity of phospho-specific antibodies.
Detection methods: Enhanced chemiluminescence with sensitive substrates or fluorescent secondary antibodies often provides better quantification of phosphorylation signals.
Remember that LTK's ER localization means its activation mechanisms likely differ from typical cell surface receptor tyrosine kinases .
LTK regulates ER-to-Golgi trafficking through modulation of ER exit sites (ERESs) . To study this function:
Combined knockdown and imaging approach:
Pharmacological inhibition:
Trafficking assays:
Implement cargo trafficking assays (e.g., VSVG-GFP or other reporter systems)
Compare trafficking kinetics between control and LTK-depleted/inhibited cells
Combine with immunofluorescence using anti-LTK antibodies to correlate LTK levels with trafficking efficiency
Co-localization studies:
These methodologies can be combined with live-cell imaging approaches for dynamic studies of trafficking regulation by LTK.
Studying LTK in tissue samples presents several challenges not encountered in cell line research:
When working with neuronal tissues, specific staining of LTK is typically localized to neurons, which can serve as an internal validation of staining specificity .
When encountering weak or non-specific LTK signals, implement these methodological solutions:
Sample preparation optimization:
Use RIPA or stronger lysis buffers with protease inhibitors
For membrane proteins like LTK, consider specialized membrane protein extraction buffers
Include proper controls (LTK-overexpressing cells, LTK knockdown samples)
Antibody selection and optimization:
Blotting conditions:
Detection enhancement:
Use more sensitive ECL substrates
Consider longer exposure times
Try signal enhancers like protein-free blocking buffers
Membrane selection:
Remember that available antibodies may not consistently detect endogenous LTK by immunoblotting, so overexpression systems may be necessary for some experiments .
When investigating LTK's role in ER stress responses, include these essential controls:
Positive controls for ER stress induction:
Genetic controls:
LTK knockdown cells (using validated siRNAs)
LTK-overexpressing cells
Rescue experiments with wild-type vs. kinase-dead LTK
Pharmacological controls:
LTK inhibitors (alectinib, crizotinib)
Control compounds with similar structures but no LTK inhibitory activity
ER stress markers to monitor:
Temporal controls:
Monitor stress markers at multiple time points
Distinguish between acute and chronic effects
Previous research has indicated that LTK inhibition increases the ER stress response (measured by XBP1s levels) in cells treated with thapsigargin, suggesting LTK may help cells cope with proteostatic challenges .
To assess and prevent cross-reactivity between LTK and ALK antibodies:
Cell line selection:
Knockdown verification:
Perform siRNA knockdown of LTK and test if antibody signal decreases
Test antibody signal in ALK knockdown cells (should remain unchanged if specific for LTK)
Epitope analysis:
Recombinant protein controls:
Test antibodies against purified recombinant LTK and ALK proteins
Perform competitive binding assays with these proteins
Known differential properties:
These approaches will help ensure the specificity of your antibody for LTK-focused research.
For successful LTK immunoprecipitation and phosphorylation analysis:
Lysis buffer composition:
Use NP-40 or RIPA buffer with phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate)
Include protease inhibitor cocktail
Buffer pH should be maintained at 7.4-7.6
Antibody selection:
Choose antibodies validated for immunoprecipitation
Antigen affinity-purified antibodies generally work best
For tagged LTK, anti-tag antibodies (e.g., anti-FLAG) often provide cleaner results
Precipitation protocol:
Washing conditions:
Use stringent washes to reduce background
Include phosphatase inhibitors in all wash buffers
Maintain cold temperature throughout to preserve phosphorylation
Elution methods:
Detection strategies:
Probe blots with phospho-specific antibodies
Consider using anti-phosphotyrosine antibodies (4G10, pY100) to detect general tyrosine phosphorylation
For comprehensive phosphosite mapping, use phospho-enrichment followed by mass spectrometry
Remember that LTK undergoes dimerization and trimerization under certain circumstances, which may affect immunoprecipitation efficiency .
LTK antibodies are valuable tools for investigating its role in disease models, particularly in systemic lupus erythematosus (SLE) and cancer:
Systemic Lupus Erythematosus (SLE) research:
Immunohistochemistry of lymphoid tissues from SLE patients/models to quantify LTK expression levels
Western blot analysis comparing LTK levels in normal vs. SLE B cells and plasma cells
Immunoprecipitation followed by phosphorylation analysis to assess activation status
Research focus: Gain-of-function LTK mutations have been observed in SLE patients and mice
Hypothesis testing: LTK overactivation may confer selective advantages to autoimmune plasma cells by enhancing secretory capacity
Cancer research applications:
Tissue microarray screening with anti-LTK antibodies to identify cancer types with altered LTK expression
Correlation of LTK levels with cancer progression and patient outcomes
Dual staining with ER stress markers to evaluate proteostatic stress
Investigation rationale: Cancer cells are considered "addicted" to secretion due to high proteostatic challenges, making LTK a potential therapeutic target
Drug response monitoring: Use LTK antibodies to assess pathway inhibition after treatment with LTK inhibitors (alectinib, crizotinib)
Methodological approaches for both disease models:
Knockdown/overexpression studies in relevant cell types
Pharmacological inhibition followed by phenotypic analysis
Phospho-specific antibody staining to assess activation status
Co-immunoprecipitation to identify disease-specific binding partners
The localization of LTK to the ER provides a unique opportunity to target ER-based signaling pathways in these disease contexts .
Adapting LTK antibody-based assays for high-content screening requires several methodological considerations:
Assay optimization:
Signal detection strategies:
Automation adaptations:
Implement automated liquid handling for consistent antibody application
Develop computational image analysis pipelines for:
Quantifying LTK expression levels
Assessing subcellular localization
Measuring co-localization with markers of interest
Controls and normalization:
Include on-plate controls (positive, negative, gradient standards)
Use nuclear or cytoplasmic markers for cell segmentation and normalization
Consider internal standards for plate-to-plate comparison
Validation of hits:
Confirm screening results with orthogonal methods (Western blot, qPCR)
Use siRNA knockdown controls to validate antibody specificity in the screening format
For LTK inhibitor screens, include known inhibitors (alectinib, crizotinib) as benchmarks
Endpoint selection:
These methodological adaptations will enable reliable high-throughput screening using LTK antibodies in various experimental contexts.
Combining proximity labeling with LTK antibodies offers powerful approaches to map the LTK interactome:
BioID/TurboID methodology:
Generate LTK-BioID fusion constructs (ensuring ER localization is maintained)
Express in relevant cell types (e.g., HepG2, lymphocytes, neurons)
Induce biotinylation of proximal proteins
Purify biotinylated proteins using streptavidin
Verify results with LTK antibodies in co-immunoprecipitation experiments
Focus on ER-localization: Since LTK is ER-resident, proximity labeling will help identify ER-specific interactors
APEX2-based approaches:
Create LTK-APEX2 fusions
Trigger brief biotinylation (1 minute) to capture transient interactions
Particularly useful for mapping dynamic interactions during ER stress
Validate findings with traditional co-immunoprecipitation using LTK antibodies
Split-BioID systems:
Validation and analysis strategies:
Confirm proximity labeling findings with traditional antibody-based co-immunoprecipitation
Use ERES-specific markers (e.g., Sec31) to validate the 10% of cells showing LTK-ERES co-localization
Employ knockdown studies of identified interactors to assess functional relevance to COPII-dependent trafficking
Comparative interactomics:
Compare LTK interactome under normal conditions versus:
ER stress conditions (thapsigargin treatment)
LTK inhibitor treatment (alectinib, crizotinib)
Disease models (SLE, cancer cell lines)
This integrated approach will provide unprecedented insights into LTK's protein interaction network within the ER environment.
Advanced approaches for studying LTK post-translational modifications include:
Phospho-specific antibody development and application:
Generate antibodies against specific phosphorylated tyrosine residues in LTK
Map the temporal sequence of tyrosine phosphorylation events
Correlate specific phosphorylation sites with downstream pathway activation
Focus on the multiple phosphotyrosines in the cytoplasmic region (aa 450-864) that interact with downstream signaling molecules
Mass spectrometry-guided antibody approaches:
Use phosphoproteomics to identify novel PTM sites on LTK
Develop and validate site-specific antibodies
Apply these antibodies to:
Track modification dynamics during ER stress
Monitor changes during inhibitor treatment
Compare normal vs. disease states
Targeted protein engineering combined with antibody detection:
Proximity ligation assays (PLA):
Combine antibodies against LTK and potential binding partners
Use PLA to visualize and quantify specific interactions
Apply in situ to tissues and cells to maintain native context
Particularly valuable for studying how PTMs alter protein interactions
Integrative approaches:
Correlate PTM patterns with:
LTK enzymatic activity
Binding partner preferences
Subcellular localization
Degradation kinetics
These methodologies provide a comprehensive toolkit for understanding how post-translational modifications regulate LTK function in normal physiology and disease states.
Single-cell analysis with LTK antibodies can reveal important functional heterogeneity:
Single-cell immunofluorescence microscopy:
Quantify LTK expression levels in individual cells
Examine subcellular distribution patterns
Correlate with ER morphology and ERES formation
Investigate the observation that approximately 10% of cells show weak co-localization between LTK and the ERES marker Sec31
Determine if this represents a distinct functional subpopulation
Flow cytometry and cell sorting:
Optimize intracellular staining protocols for LTK detection
Sort cells based on LTK expression levels
Characterize sorted populations for:
Secretory capacity
ER stress responses
Sensitivity to LTK inhibitors
Single-cell Western blotting:
Adapt LTK antibody protocols for microfluidic Western platforms
Compare LTK expression and phosphorylation states at single-cell resolution
Correlate with cellular phenotypes
Imaging mass cytometry:
Correlation with single-cell transcriptomics:
Integrate antibody-based protein detection with scRNA-seq data
Identify transcriptional signatures associated with varying LTK expression
Investigate post-transcriptional regulation by comparing mRNA and protein levels
This multi-modal single-cell approach will provide unprecedented insights into the functional significance of cell-to-cell variability in LTK expression, potentially revealing specialized subpopulations with distinct roles in secretion regulation and ER homeostasis.
LTK antibodies can facilitate the development of ER proteostasis-targeted therapeutics through:
Target validation strategies:
Use antibodies to correlate LTK expression/activation with disease severity
Perform IHC studies across tissue microarrays of various diseases with ER stress components
Validate LTK as a drug target based on its role as the first identified ER-resident receptor tyrosine kinase regulating COPII-dependent trafficking
Therapeutic antibody development:
Although LTK is primarily ER-localized, investigate potential exofacial epitopes for therapeutic antibody targeting
Explore antibody-drug conjugates targeted to cells overexpressing LTK
Develop cell-penetrating antibodies or antibody fragments to reach intracellular LTK
Small molecule drug discovery support:
Implement antibody-based assays to screen for compounds that modulate LTK activity
Use phospho-specific antibodies to monitor target engagement
Develop competitive binding assays to identify compounds that disrupt specific protein-protein interactions
Build on existing knowledge of LTK inhibitors (alectinib, crizotinib) that reduce ERES numbers
Disease-specific applications:
Systemic lupus erythematosus: Investigate how gain-of-function LTK mutations affect ER proteostasis in plasma cells
Cancer: Explore LTK inhibition as a strategy to induce ER stress in cancer cells already challenged by proteostatic stress
Neurodegenerative diseases: Study LTK's role in neuronal ER homeostasis, given its expression in cerebral cortex neurons
Combination therapy development:
These approaches position LTK antibodies as crucial tools in developing a new class of therapeutics targeting ER proteostasis regulation.
Advanced methodologies for studying LTK dynamics during ER stress include:
Biosensor development:
Design FRET-based biosensors to monitor LTK conformational changes
Develop activity reporters based on known LTK substrates
Create tension sensors to detect mechanical forces during ER membrane remodeling
Validate these tools using existing antibodies as references
Live-cell super-resolution microscopy:
Conditional protein regulation:
Microfluidic approaches:
Design chambers for controlled application/removal of ER stressors
Combine with live-cell antibody-based detection methods
Monitor LTK expression, localization, and activation in real-time during stress induction and recovery
Spatial transcriptomics integration:
These methodological innovations will provide unprecedented insights into the temporal dynamics of LTK function during ER stress, potentially revealing new therapeutic windows for intervention in diseases characterized by ER dysfunction.
Cross-species LTK antibody studies can reveal evolutionary aspects of ER signaling:
Epitope conservation analysis:
Functional conservation studies:
Tissue expression pattern comparison:
Disease model translation:
Developmental regulation:
Map LTK expression during embryonic development across species
Identify conserved vs. divergent temporal expression patterns
Link to the development of secretory tissues
This evolutionary perspective will provide valuable insights into the fundamental importance of LTK-mediated ER signaling and help distinguish conserved core functions from species-specific adaptations.
Understanding these comparative strengths and limitations enables selection of the optimal imaging approach for specific LTK research questions.
Each approach offers distinct advantages for investigating different aspects of LTK biology, from protein interactions to signaling pathway regulation.